Next Article in Journal
Isolation and Characterization of Streptococcus mutans Phage as a Possible Treatment Agent for Caries
Previous Article in Journal
Complement Proteins as Soluble Pattern Recognition Receptors for Pathogenic Viruses
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Article

Scutellaria barbata D. Don Inhibits the Main Proteases (Mpro and TMPRSS2) of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection

1
School of Chinese Medicine, China Medical University, Taichung 40402, Taiwan
2
Department of Chinese Medicine, China Medical University Hospital, Taichung 40402, Taiwan
3
Research Cancer Center for Traditional Chinese Medicine, Department of Medical Research, China Medical University Hospital, Taichung 40402, Taiwan
4
An-Nan Hospital, China Medical University, Tainan 709, Taiwan
5
Research Center for Cancer Biology, China Medical University, Taichung 40402, Taiwan
6
New Drug Development Center, China Medical University, Taichung 40402, Taiwan
7
Graduate Institute of New Drug Development, China Medical University, Taichung 40402, Taiwan
8
Center for Molecular Medicine, China Medical University Hospital, Taichung 40402, Taiwan
9
Department of Biological Science and Technology, China Medical University, Taichung 40402, Taiwan
10
Graduate Institute of Biomedical Sciences, China Medical University Taichung 40402, Taiwan
11
Department of Biotechnology, Asia University, Taichung 41354, Taiwan
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Viruses 2021, 13(5), 826; https://doi.org/10.3390/v13050826
Submission received: 31 March 2021 / Revised: 28 April 2021 / Accepted: 29 April 2021 / Published: 2 May 2021
(This article belongs to the Topic Broad-Spectrum Antiviral Agents)

Abstract

:
In late 2019, the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) pandemic emerged to severely impact the global population, creating an unprecedented need for effective treatments. This study aims to investigate the potential of Scutellaria barbata D. Don (SB) as a treatment for SARS-CoV-2 infection through the inhibition of the proteases playing important functions in the infection by SARS-CoV-2. FRET assay was applied to investigate the inhibitory effects of SB on the two proteases involved in SARS-CoV-2 infection, Mpro and TMPRSS2. Additionally, to measure the potential effectiveness of SB treatment on infection inhibition, cellular models based on the Calu3 and VeroE6 cells and their TMPRSS2- expressing derivatives were assessed by viral pseudoparticles (Vpp) infection assays. The experimental approaches were conjugated with LC/MS analyses of the aqueous extracts of SB to identify the major constituent compounds, followed by a literature review to determine the potential active components of the inhibitory effects on protease activities. Our results showed that SB extracts inhibited the enzyme activities of Mpro and TMPRSS2. Furthermore, SB extracts effectively inhibited SARS-CoV-2 Vpp infection through a TMPRSS2-dependent mechanism. The aqueous extract analysis identified six major constituent compounds present in SB. Some of them have been known associated with inhibitory activities of TMPRSS2 or Mpro. Thus, SB may effectively prevent SARS-CoV-2 infection and replication through inhibiting Mpro and TMPRSS2 protease activities.

1. Introduction

Emerging in late 2019, the spread of severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), named coronavirus disease-2019 (COVID-19) by the World Health Organization (WHO), rapidly reached global pandemic proportions. By late 2020, the total number of confirmed COVID-19 cases worldwide had reached over 72 million, with total reported deaths of more than 1.6 million [1]. The pandemic outbreak has severely impacted governments and health care systems around the world [2,3]. However, there currently exist only supportive therapeutic strategies to manage COVID-19 infection [4], while targeted antiviral drugs have yet to be developed [5]. As an ancient therapeutic strategy, traditional Chinese medicine (TCM) has historically played an important role in the treatment and prevention of diseases [6,7].
Scutellaria barbata D. Don (SB), also known as “Ban Zhi Lian”, is a widely used herb in TCM, demonstrating heat-clearing and detoxifying properties [8]. Neo-clerodane diterpenoids and flavonoids are the main compounds of SB, followed by polysaccharides, volatile oils, and steroids. In recent decades, studies of SB have focused on its anticancer, antioxidant, anti-inflammatory, antiviral, and antibacterial activities [8,9]. Most research into the cytotoxic effects (anti-tumor effects) of neo-clerodane diterpenoids, both in vitro and in vivo, have been related to hepatic cancer, breast cancer, colorectal cancer, lung cancer, and ovarian cancer [10,11,12,13,14]. Guo et al. reported that the total flavone could prevent anti-parainfluenza viral infection, and improve the membrane fluidity of Hep-2 cells [15]. Meanwhile, Wu et al. identified 11 diterpenoids that exhibited moderate to potent inhibition activities against Epstein–Barr virus lytic replication [16]. Antibacterial effects have also been reported, wherein SB showed an inhibitory effect against the extensively drug-resistant Acinetobacter baumannii [17]; in addition, gram-positive bacteria have demonstrated sensitivity to inhibition by SB [18]. Furthermore, apigenin and luteolin isolated from SB were found to be selectively toxic to the methicillin-resistant S. aureus and methicillin-sensitive S. aureus strains [19]. Other conditions, including diabetic retinopathy [20], cognitive dysfunction [21,22], and myocardial ischemia–reperfusion injury [23] have also been shown to benefit from SB treatment.
From the beginning of the SARS-CoV-2 pandemic, scientists have been eagerly investigating the key infection routes and viral infection cycle, with the aim of developing effective targeted antiviral treatments. SARS-CoV-2 uses the angiotensin converting enzyme II (ACE2) receptor to enter the host cell, in synergy with transmembrane serine protease 2 (TMPRSS2) [24]. TMPRSS2 is a cell-surface protein located on respiratory and gastroenterology epithelial cells; importantly, the coronavirus, including SARS-CoV-2, are known to enter human cells through interaction of viral spike protein with host ACE2 then cleaved by host TMPRSS2 protease. Thus, the rapid spread of communal infection is reliant on the efficiency of the ACE2-spike interaction and TMPRSS2 protease activity [25]. Meanwhile, the TMPRSS2: ERG fusion gene has been associated with prostate cancer and serves as a biomarker for the diagnosis and stratification of androgen-sensitive prostate cancer [26]. Although earlier studies primarily focused on prostate cancer, more recent studies have investigated TMPRSS2/ACE2 integrity as related to SARS-CoV-2 infection. Several studies have indicated that modifications to the renin-angiotensin pathway or TMPRSS2 expression would break down viral infections [25,27,28].
The screening of natural compounds in several studies, including 10-hydroxyusambarensine and cryptospirolepine, has revealed the potential of inhibiting host cell entry by altering the ACE2-TMPRSS2 pathway [29,30,31]. Compounds inhibiting TMPRSS2 protease activity have notably been associated with reduced infection activity of the SARS-CoV-2 Vpp [32]. Additionally, more recent investigations have been conducted to determine effects related to Mpro, also called 3C-like protease (3CLpro), one of the SARS-CoV-2 nonstructural proteins. Mpro cleaved the original viral polyproteins—pp1a and pp1ab—with its proteolytic activity. Of note, the 33.8-kDa Mpro is necessary for viral replication and transcription [33]. Recent research has focused on developing a screening strategy for effective Mpro inhibitors, potentially associated with SARS-CoV-2 infection inhibition [34,35]. In structural based screenings, natural compounds including epigallocatechin gallate, epicatechin gallate, gallocatechin-3-gallate, glycyrrhizin, bicyclogermacrene, tryptanthrine, β-sitosterol, indirubin, indican, indigo, hesperetin, chrysophanic acid, rhein, berberine, and β-caryophyllene have been identified as potential herbal Mpro inhibitors against SARS-CoV-2 infection [36,37]. Furthermore, herbs including Glycyrrhiza, Tinospora cordifolia, Uncaria tomentosa have demonstrated Mpro inhibitory effects [38,39,40].
Herbal medicines are often considered as low-toxicity alternative treatment modalities, while various herbal compounds are regarded as potential treatments for SARS-CoV-2. Recent studies have revealed the effectiveness of several natural compounds at TMPRSS2 or Mpro inhibition [30,32,38,41]. However, few studies investigating TCM herbs have been reported. Indeed, previous studies have shown that SB (Ban-zhi-lian) exhibits notable antiviral, antibacterial, anticancer, and immunomodulation effects; although investigation into the potential anti-SARS-CoV-2 effects of SB and its role in TMPRSS2 and Mpro inhibition remains limited. Thus, this study aims to evaluate the effectiveness of SB on the inhibition of Mpro and TMPRSS2 associated with SARS-CoV-2 infection activity. In this current study, we show that SB inhibits protease activity of SARS-CoV-2 Mpro and TMPRSS2 and suppresses pseudoviral entry, paving a way for therapeutic application as an anti-SARS-CoV-2 agent.

2. Materials and Methods

2.1. Preparation of Scutellaria barbata (SB) Aqueous Extract

The SB utilized in this study is recognized in accordance with the definition described in Flora of Taiwan, with a sample of SB deposited in Set D, TN: BRCM 4616 in the herbarium of National Taiwan University. The extract was prepared from the whole plant according to Taiwanese GMP methods and guidelines, and the concentrated powder was manufactured by a recognized GMP company (Kaiser Pharmaceutical Co., Ltd., Tainan, Taiwan). Of note, it is applied in clinical practice and covered by the National Health Insurance system of Taiwan. Briefly, the 2.5 g powder was boiled with 10 mL ddH2O for 10 min, followed by centrifugation of 3000 rpm for 10 min and 12,000 rpm for 15 min. The resulting extract was filtered with 0.22 μm microfilter and then aliquot for reservation. The extraction rate was 14.2%, i.e., 1 g of concentrated powder (extract: excipient 1: 1) is equal to 3.5 g of the original herb. The stock concentration was 400 mg/mL.

2.2. Liquid Chromatography–Mass Spectrometry (LC/MS) Analysis

The aqueous extracts of SB were analyzed using a Velos Pro dual-pressure linear ion trap mass spectrometer (Thermo Fisher Scientific, San Jose, CA, USA) equipped with an Agilent 1100 Series binary high-performance liquid chromatography pump (Agilent Technologies, Palo Alto, CA, USA), and a Famos autosampler (LC Packings, San Francisco, CA, USA), using a Xbridge column (1 mm I.D. × 150 mm, 3.5 um, 130 Å). Briefly, the gradient employed was 2% buffer B at 2 min, to 98% buffer B at 20 min, with a flow rate of 50 μL/min, where buffer A was 0.1% formic acid/H20 and buffer B was 0.1% formic acid/acetonitrile. The survey scan was acquired in the mass range m/z 200–2000. Electrospray voltage was maintained at 4 kV and capillary temperature was set at 275 °C.

2.3. Estimation of the Average Molecular Weight of Aqueous Extract of SB

The mass range 200–2000 was divided into 45,000 intervals and the mass intensities in each interval were summed from 5340 scan events during a 30 min analytical process. The relative intensity ratio of each interval was acquired by the calculation of (an induvial mass intensity/total mass intensity) as shown in Supplementary Table S1.
i = 1 5340 [ ( mass ) i × ( intensity   ratio ) i ] . The average molecular weight = 547.6 Da.

2.4. Protein Sample Preparation

Recombinant SARS-CoV-2 Mpro and its protein substrate were prepared following the previously described protocol [41]. Briefly, the target gene encoding SARS-CoV-2 Mpro (residues 3264–3569, UniProt accession: P0DTD1) and the fluorescent protein substrate of Mpro (CFP-TSAVLQSGFRKM-YFP) were synthesized and subcloned into pSol SUMO vector (Lucigen) and pET16b vector, respectively. Overexpression of SARS-CoV-2 Mpro was induced by 0.5 mM IPTG until O.D.600 reached 0.6 and further incubated for 18 h at 20 °C, while its fluorescent protein substrate was induced by 20% L-rhamnose and incubated for 20 h at 16 °C. Human TMPRSS2 (amino acids 256-492, UniProt accession: C9JKZ3) with N-terminal His6-MBP-tag and its protein substrate (GFP- QTQTNSPRRARSVAS-RFP) were constructed and recombinantly expressed in E. coli BL21 (DE3) as previously described [41]. All the protein samples were purified by immobilized-metal affinity chromatography using ÄKTA™ go chromatography system (Cytiva) according to manufacturer instructions. The purity and monodispersity of protein samples were further achieved by an additional purification step using size-exclusion chromatography (HiLoad Superdex 200, GE Healthcare, Chicago, IL, USA).

2.5. Fluorescence Resonance Energy Transfer (FRET) Assay

The enzyme activity of SARS-CoV-2 Mpro was determined by FRET assay, as previously detailed [41]. For the activity assay of TMPRSS2, 100 μL reaction volume containing 25 μM TMPRSS2 protein in the presence or absence of SB was pre-incubated at room temperature for 30 min. The reaction was initiated by addition of the fluorescent protein substrate and monitored continuously for 2 h at 536 nm after excitation at 506 nm using a Synergy H1 microplate reader (BioTek Instruments, Inc., Winooski, VT, USA). The concentrations of SB used in the FRET assays of SARS-CoV-2 Mpro were from 0.5 mg/mL to 4 mg/mL with two-fold series dilution. The IC50 value of SB on inhibition of SARS-CoV-2 Mpro was determined by using 7 different concentrations of SB and calculated by Prism 9 software. The final concentration of SB used in the FRET assays of TMPRSS2 was 4 mg/mL.

2.6. Cells

Calu3 cells were cultured in Modified Eagle’s Medium (MEM) supplemented with 10% fetal bovine serum (FBS), 1 × NEAA, and 1% penicillin/streptomycin. VeroE6 were cultured in Dulbecco’s Modified Eagle’s Medium (DMEM) supplemented with 10% FBS, 1× GlutaMAX, and 1% penicillin/streptomycin. TMPRSS2 expressing cells of Calu3 and VeroE6 cell lines were generated by transfection of the pCMV3-TMPRSS2-Flag plasmid and selection by hygromycin.

2.7. Viral Pseudoparticles (Vpp) Infection and Inhibition

For the infection experiments, 10,000 cells were seeded in 96-well plates. The cells were then pre-incubated with a medium containing different concentrations of SB or H2O (vehicle control) for 1 h at 37 °C and 5% CO2, before they were infected with the Vpp harboring SARS-CoV-2-S and luciferase reporter (purchased from National RNAi Core Facility (NRC), Academia Sinica, Taipei, Taiwan), followed by centrifugation at 1250 g for 30 min. After 24 h incubation, the Cell Counting Kit-8 (CCK-8) assay (Dojindo Laboratories, Mashiki, Kumamoto, Japan) was performed to measure cell viability. Then, each sample was mixed with an equal volume of ready-to-use luciferase substrate Bright-Glo Luciferase Assay System (Promega, Madison, WI, USA), with luminescence measured immediately by the GloMax Navigator System (Promega, Madison, WI, USA). Raw luminescence values, indicating luciferase activity, were recorded as counts per second. The relative light unit (RLU) was normalized with cell viability first, then the control group was set as 100%, and the relative infection efficiencies were calculated.

2.8. Statistical Analysis

All statistical analyses were calculated with SigmaStat statistical software (version 2.0, Jandel Scientific, SanRafael, CA, USA). All data originating from experiments were performed in triplicate and represented as mean ± standard deviation (SD). Statistical significance was calculated using student t-test. ANOVA was performed when multiple comparisons were evaluated. P values less than 0.05 were considered to be significant. All experiments were repeated at least three times independently.

3. Results

3.1. Initial Screening of Scutellaria barbata against SARS-CoV-2 Mpro and TMPRSS2

For the initial screening, we selected twenty herbs in two independent batches to identify protease enzyme activities against SARS-CoV-2 Mpro. One of these herbs, Scutellaria barbata (Ban-zhi-lian), which is utilized in treating liver, lung, and colon cancer [42,43,44], was determined the best candidate for SARS-CoV-2 inhibition. We, therefore, investigated the aqueous extracts of SB on the inhibition of the main protease activity of SARS-CoV-2, a primary therapeutic target due to its critical role in viral replication. The FRET assay revealed that 4 mg/mL SB effectively inhibited the SARS-CoV-2 Mpro, with a remarkable inhibition rate of 93.3% (Figure 1). To further evaluate the efficacy of the aqueous extracts of SB, we performed a dose–response titration enzymatic assay. The results showed that 2 mg/mL, 1 mg/mL, and 0.5 mg/mL SB inhibited the protease activity of SARS-CoV-2 Mpro at rates of 79.5%, 30.1%, and 1.6%, respectively (Figure 1). The half-maximal inhibitory concentration (IC50) of SB on inhibition of SARS-CoV-2 Mpro was further determined to be 1.27 mg/mL (calculated by using the average molecular weight of 547.6 Da of SB) (Figure 2). Another important therapeutic target to combat SARS-CoV-2 is TMPRSS2, which plays an essential role in the cellular entry of SARS-CoV-2. The in vitro enzymatic assay demonstrated that 4 mg/mL SB had a moderate inhibitory effect (54.8%) on TMPRSS2 activity (Figure 3).

3.2. Scutellaria barbata Inhibited Pseudovirus Infection through TMPRSS2

SARS-CoV-2 can enter the host cell via both endosomal and non-endosomal pathways. The endosomal-mediated pathway includes the S protein binding to ACE2 and subsequent dynamin/clathrin-mediated internalization of endosomal vesicles. In addition to the endosomal-mediated pathway, host TMPRSS2 also play critical roles in the non-endosomal entry of SARS-CoV-2 [1]. VeroE6 cell line (monkey kidney epithelial cell line) is used as a standard model for endosomal entry pathway, because this cell line presents relatively high ACE2 expression and low TMPRSS2 expression. Calu3 cell line (human lung cancer cell line), as with the airway epithelial cells, express high amounts of TMPRSS2 and SARS-CoV-2 entry into these cells is dependent on TMPRSS2 [45]. Calu3 and VeroE6 cells with and without TMPRSS2 over-expression were treated with Scutellaria barbata. After 24 h treatment, the cell viability measured by CCK8 assay demonstrated no cytotoxicity for both (Supplementary Figure S1). Our results already demonstrated that SB could potently inhibit TMPRSS2 enzyme activity in vitro (Figure 3). To further investigate whether SB would block SARS-CoV-2 infection through inhibiting TMPRSS2 activity, we first generated the TMPRSS2-expressing cells in both VeroE6 and Calu3 cells. Then, these cells were pre-treated for 1 h with 4 or 8 mg/mL of SB and then infected with the SARS-CoV-2 Vpp and infection efficiency was determined according to luciferase activities after 24 h of infection. The results demonstrated that SB blocked infection of the SARS-CoV-2 Vpp in Calu3 cells, but not in VeroE6 cells (Figure 4). Furthermore, SB showed a higher inhibitory effect in both over-expressing TMPRSS2 cells (Figure 4). These results indicate that SB can inhibit SARS-CoV-2 entry by affecting TMPRSS2 activity.

3.3. The Scutellaria barbata Fingerprint

To analyze the composition of SB, the aqueous extracts of SB were separated by HPLC (high performance liquid chromatography), and the exact molecular weights of the individual constituent compounds were determined by mass spectrometry. The major ingredients identified, including apigenin, naringenin, scutellarin, baicalein, luteolin, and wogonin are consistent with previous reports [8,46], as shown in Figure 5. It is interesting to note that several derivatives of SB, including scutellarin, baicalein, wogonin, and luteolin have also been reported to inhibit Mpro and TMPRSS2 (please see discussion), which provides a scientific basis for the SB to inhibit SARS-CoV-2 infection.

4. Discussion

The main protease (Mpro) functions in a dimeric form, involving in proteolytic processing of replicase polyproteins in SARS-CoV-2. Drugs targeting Mpro with broad-spectrum antiviral activity and lower the risk of mutation-mediated drug resistance are potent antiviral agents against SARS-CoV-2 infection [47]. Based on Mpro inhibitors of TCM herbs screening, Zingiberis Rhizoma Recens, Asteris Radix et Rhizoma, Notoginseng Radix et Rhizoma, Chuanxiong Rhizoma, Salviae Miltiorrhizae Radix et Rhizoma, Zingiberis Rhizoma, Dianthi Herba, Rhei Radix et Rhizoma, Cistanches Herba are reported by Ma et al. [48]. In this study, we found that SB effectively inhibited the SARS-CoV-2 Mpro. Additionally, Xia et al. found that Shufeng Jiedu capsules can reduce the activity of NFκB in the lungs of HCoV-229E mice. Its active components, wogonin, quercetin, and polydatin, bind directly to the SARS-CoV-2 Mpro [49].
Spike-ACE2 interaction and TMPRSS2 protease activity are critical for host cell-mediated viral entry of SARS-CoV-2. Thus, drugs which inhibit TMPRSS2 or spike-ACE2 interaction, thereby blocking viral fusion, are of particular interest in the search for SARS-CoV-2 treatments. Various existing drugs, including chloroquine, hydroxychloroquine, and other antiviral drugs such as remdesivir (a nucleotide analogue for Ebola virus), lopinavir and ritonavir (HIV protease inhibitors), and arbidol and favipiravir (broad-spectrum anti-influenza drugs) were early potential treatment candidates for SARS-CoV-2 [50]. Meanwhile, bromhexine hydrochloride is a mucolytic cough suppressant, and as a TMPRSS2 inhibitor, may hold potential for treatment of SARS-CoV-2 infection [51]. Another drug, camostat, which is used to treat chronic pancreatitis, could inhibit SARS-CoV-2 infection via serine protease effects [52]. Notably, 10–hydroxyusambarensine, cryptospirolepine, and cryptoquindoline, which are extracted from African medicinal plants, are reported to exhibit superior TMPRSS2 inhibitory effects [31]. Of those three compounds, 10–hydroxyusambarensine was identified as the most effective, by docking into the S1-specificity pocket of TMPRSS2 [31]. Other natural compounds, including geranium, lemon essential oils, homoharringtonine, and halofuginone have also demonstrated TMPRSS2 inhibitory effects [30,53]. Of note, several derivatives of SB, including scutellarin, baicalein, wogonin, and luteolin have also been reported to inhibit Mpro and TMPRSS2, further supporting the promising results reported herein. Moreover, a recent study indicated another host protease, furin, is also involved in virus infection via a separate mechanism [54], although it is currently unclear whether SB may also exert an effect on a furin-mediated pathway.
This is the first study to report on the role of SB against SARS-CoV-2 infection through the inhibition of the main protease. The FRET assay demonstrated that SB blocked SARS-CoV-2 infection through inhibition of Mpro and TMPRSS2. Furthermore, in Calu3 cells and over-expressing TMPRSS2 cells, SB treatments inhibited the SARS-CoV-2 Vpp infection. These results indicate the potential of SB as an alternative drug to treat SARS-CoV-2 infection. Several compounds present in SB have recently been recognized for their potential benefits in clinical application. As a major compound of SB, scutellarin has exhibited NLRP3 inflammasome suppression, thereby rescuing mice from bacterial sepsis [55]. In addition, a silico screening report of scutellarin revealed Mpro inhibitory effects, supporting our study results [56]. Meanwhile, baicalein, which is a flavone extracted from the roots of Scutellaria baicalensis and Scutellaria lateriflora, has exhibited notable anti-oxidant, anti-virus, anti-bacteria, anti-inflammatory, anti-cancer, and immunoregulatory effects [57,58,59]. Huang et al. have reported that natural compounds including baicalein, luteolin, naringenin, and wogonin suppress ACE2 and Mpro protease activity to suppress SARS-CoV-2 [60]. Baicalein, a main ingredient of the TCM herbal formula Shuanghuanglian, has demonstrated anti-viral effects through inhibiting Mpro activity [61,62]. Additionally, studies have reported that baicalein significantly affects both ACE2 and TMPRSS2 [63,64]. A notable screening study, suggesting the compound as a candidate for the treatment of SARS-CoV-2, reported that baicalein also binds with the SARS-CoV-2 nonstructural protein 14 (NSP14) [65]. Another constituent compound of SB is luteolin, a yellow dye compound that is obtained primarily from Reseda luteola, is in fact an ingredient in many foods, including celery, broccoli, parsley, chamomile tea, carrots, olive oil, peppermint, oranges, and medicinal herbs such as Artemisia. Luteolin has been shown to exhibit anti-inflammatory, anti-apoptosis, and anti-cancer effects [66,67,68]. Furthermore, luteolin, one of the major compounds in Lianhua Qingewen capsules and Maxingyigan, has been reported as a potential treatment candidate of SARS-CoV-2 via targeting of the Akt1 (serine/threonine kinase 1) pathway [69]. Liang et al. also demonstrated that luteolin could inhibit SARS-CoV-2 by interleukin-6 down-regulation [70]. Luteolin has been associated with anti-coronavirus effects via both inhibiting ACE2 and Mpro activity, further confirming our results [71,72,73]. Another constituent component isolated from SB, wogonin, is a key flavonoid for quality control and exhibits anti-inflammatory, anti-cancer, anti-viral, anti-angiogenesis, anti-oxidant, and neuro-protective effects [8]. Together with phytol, luteolin, and hispidulin, wogonin is one of the active constituents in the chloroform fraction of SB, which has demonstrated dose-dependent cytotoxicity on six cancer cell lines [74]. Wogonin presents anti-viral activities against influenza infection via modulation of AMP-activated protein kinase (AMPK) pathways, and anti-fibrotic effects in liver fibrosis via regulating the activation and apoptosis of hepatic stellate cells [75]. Thus, wogonin may present a therapeutic option to attenuate the cytokine storm in acute symptomatic SARS-CoV-2 infections due to the immunomodulatory response [76] and its anti-fibrotic effects in the convalescence period [75]. Yet another flavonoid compound for quality control in SB [8], naringenin, can inhibit two-pore channels (TPCs) [77,78], which regulate the trafficking of the virus to late-endosomes/lysosomes following entry into cells [79]. Several studies have investigated and reported on the antiviral activities of naringenin against hepatitis C (HCV), influenza A, Zika, and Dengue Fever [80,81,82,83]. Thus, it is reasonable to suggest that naringenin may also be effective in combating SARS-CoV-2. Finally, the compound apigenin has been shown to suppress influenza A virus-induced RIG-I activation and viral replication [84], and EBV reactivation [85]. It also exhibits an inhibitory effect on the foot-and-mouth disease virus [86], the African swine fever virus [87], the buffalopox virus [88], and the enterovirus [73,89]. However, it must be noted that in order to reach effective plasma concentrations. There is a potential limitation to take a relatively high dose of SB in vivo. Nevertheless, taken together, SB and its major constituent components indeed demonstrate various anti-viral effects which thus warrant consideration for inclusion in the development of future therapeutic strategies for SARS-CoV-2 infection.

5. Conclusions

SARS-CoV-2 is an RNA-virus currently challenging health care systems worldwide. As candidates for SARS-CoV-2 prevention and treatment, anti-viral herbal compounds present low-cost and low-toxicity options. The anti-viral, anti-bacterial, anti-inflammation, and anti-cancer effects of SB indicate the therapeutic promise of SB. This is the first study to report that SB effectively inhibits Mpro and TMPRSS2 activity in vitro. Inhibition of TMPRSS2 is able to suppress SARS-CoV-2 virus particle, Vpp to infect host cells. And suppression of Mpro activity is known to inhibit virus replication. Thus, SB may associate with dual activities to inhibit both SARS-CoV-2 virus infection and replication. Our results demonstrate that SB would be an ideal candidate for inclusion in the development of SARS-CoV-2 treatments. Further investigation into the clinical efficacy of SB associated with its distinct molecular mechanisms is thus warranted.

Supplementary Materials

The following are available online at https://www.mdpi.com/article/10.3390/v13050826/s1, Figure S1: Scutellariabarbatashowed no cytotoxicity. Calu3 and VeroE6 cells with and without TMPRSS2 over-expression were treated withScutellariabarbata. Table S1: Scutellaria barbata (SB).

Author Contributions

S.-T.H. wrote the paper and unify the entire study. Y.C., W.-C.C., H.-F.C., H.-C.L., and Y.-C.L. wrote the paper and amended the references. Y.C., H.-F.C., W.-J.W., Y.-C.W., and C.-S.Y. offered technique assistance, performed the experiments, and analyzed the data. W.-C.C. analyzed the fingerprint and confirmed the structures of extract. S.-C.W. offered technique assistance and reviewed the text. M.-C.H. designed, conceived the study, and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by the Ministry of Science and Technology of Taiwan (MOST 109-2327-B-039-003 (M.-C.H.)) and funded by China Medical University Hospital (DMR-109-199, DMR-110-004, DMR-110-009 and CRS-108-001 (S.-T.H)), and the Chinese Medicine Research Center, China Medical University, under the Higher Education Sprout Project, Ministry of Education (CMRC-CHM-1 (S.-T.H)) in Taiwan.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The data presented in this study are available in this article and on request from the author and corresponding author.

Acknowledgments

The Calu3 and VeroE6 cell lines are generous gifts from Chien-Te (Kent) Tseng (University of Texas Medical Branch in Galveston, Texas) and Sui-Yuan Chang (National Taiwan University, Taiwan), respectively. The authors would like to thank James Waddell of Concise Language Services for the critical reading and revision of our manuscript.

Conflicts of Interest

The authors declare that they have no competing interests.

Abbreviations

The following abbreviations are used in this manuscript:
3CLpro3C-like protease
ACE2angiotensin converting enzyme II
Akt1serine/threonine kinase 1
AMPKAMP-activated protein kinase
Calu-3human bronchial epithelial cells
CCK-8cell counting kit-8
COVID-19coronavirus disease-2019
DMEMDulbecco’s Modified Eagle’s Medium
FBSfetal bovine serum
FRETfluorescence resonance energy transfer
HPLC high performance liquid chromatography
MEMModified Eagle’s Medium
MproMain protease
SARS-CoV-2severe acute respiratory syndrome coronavirus 2
SBScutellaria barbata D. Don
SDstandard deviation
TCMtraditional Chinese medicine
TMPRSS2transmembrane serine protease 2
VeroE6monkey kidney epithelial cells
Vppviral pseudoparticles
WHOWorld Health Organization

References

  1. World Health Organization. Coronavirus Disease (COVID-2019) Situation Reports. Available online: https://www.who.int/publications/m/item/weekly-epidemiological-update---22-december-2020 (accessed on 22 December 2020).
  2. Fernandes, N. Economic effects of coronavirus outbreak (COVID-19) on the world economy. SSRN Electron. J. 2020. [Google Scholar] [CrossRef]
  3. Nicola, M.; Alsafi, Z.; Sohrabi, C.; Kerwan, A.; Al-Jabir, A.; Iosifidis, C.; Agha, M.; Agha, R. The socio-economic implications of the coronavirus pandemic (COVID-19): A review. Int. J. Surg. 2020, 78, 185–193. [Google Scholar] [CrossRef] [PubMed]
  4. World Health Organization. Clinical Management of Severe Acute Respiratory Infection When COVID-19 Is Suspected. Available online: https://www.who.int/publications/i/item/10665-332299 (accessed on 30 April 2021).
  5. Zumla, A.; Chan, J.F.W.; Azhar, E.I.; Hui, D.S.C.; Yuen, K.-Y. Coronaviruses—Drug discovery and therapeutic options. Nat. Rev. Drug Discov. 2016, 15, 327–347. [Google Scholar] [CrossRef] [Green Version]
  6. Hanson, M. Speaking of Epidemics in Chinese Medicine Disease and the Geographic Imagination in Late Imperial China, 1st ed.; Routledge: New York, NY, USA, 2011; p. xi-265. [Google Scholar]
  7. Huang, S.T.; Lai, H.C.; Lin, Y.C.; Huang, W.T.; Hung, H.H.; Ou, S.C.; Lin, H.J.; Hung, M.C. Principles and treatment strategies for the use of Chinese herbal medicine in patients at different stages of coronavirus infection. Am. J. Cancer Res. 2020, 10, 2010–2031. [Google Scholar]
  8. Wang, L.; Chen, W.; Li, M.; Zhang, F.; Chen, K.; Chen, W. A review of the ethnopharmacology, phytochemistry, pharmacology, and quality control of Scutellaria barbata D. Don. J. Ethnopharmacol. 2020, 254, 112260. [Google Scholar] [CrossRef] [PubMed]
  9. Chen, Q.; Rahman, K.; Wang, S.J.; Zhou, S.; Zhang, H. Scutellaria barbata: A review on chemical constituents, pharmacological activities and clinical applications. Curr. Pharm. Des. 2020, 26, 160–175. [Google Scholar] [CrossRef] [PubMed]
  10. Kan, X.; Zhang, W.; You, R.; Niu, Y.; Guo, J.; Xue, J. Scutellaria barbata D. Don extract inhibits the tumor growth through down-regulating of Treg cells and manipulating Th1/Th17 immune response in hepatoma H22-bearing mice. BMC Complement. Altern. Med. 2017, 17, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  11. Wang, M.; Ma, C.; Chen, Y.; Li, X.; Chen, J. Cytotoxic Neo-Clerodane Diterpenoids from Scutellaria barbata D.Don. Chem. Biodivers. 2019, 16, e1800499. [Google Scholar] [CrossRef] [PubMed]
  12. Chen, C.C.; Kao, C.P.; Chiu, M.M.; Wang, S.H. The anti-cancer effects and mechanisms of Scutellaria barbata D. Don on CL1–5 lung cancer cells. Oncotarget 2017, 8, 109340–109357. [Google Scholar] [CrossRef] [PubMed]
  13. Shiau, A.L.; Shen, Y.T.; Hsieh, J.L.; Wu, C.L.; Lee, C.H. Scutellaria barbata inhibits angiogenesis through downregulation of HIF-1 α in lung tumor. Environ. Toxicol. 2014, 29, 363–370. [Google Scholar] [CrossRef] [PubMed]
  14. Zhang, L.; Ren, B.; Zhang, J.; Liu, L.; Liu, J.; Jiang, G.; Li, M.; Ding, Y.; Li, W. Anti-tumor effect of Scutellaria barbata D. Don extracts on ovarian cancer and its phytochemicals characterisation. J. Ethnopharmacol. 2017, 206, 184–192. [Google Scholar] [CrossRef] [PubMed]
  15. Guo, S.S.; Shi, Y.J.; Gao, Y.J.; Su, D.; Cui, X.L. The cytology mechanism of anti-parainfluenza virus infection of total flavone of Scutellaria barbata. Yao Xue Xue Bao Acta Pharm. Sin. 2009, 44, 1348–1352. [Google Scholar]
  16. Wu, T.; Wang, Q.; Jiang, C.; Morris-Natschke, S.L.; Cui, H.; Wang, Y.; Yan, Y.; Xu, J.; Lee, K.H.; Gu, Q. Neo-clerodane diterpenoids from Scutellaria barbata with activity against Epstein-Barr virus lytic replication. J. Nat. Prod. 2015, 78, 500–509. [Google Scholar] [CrossRef] [PubMed]
  17. Tsai, C.C.; Lin, C.S.; Hsu, C.R.; Chang, C.M.; Chang, I.W.; Lin, L.W.; Hung, C.H.; Wang, J.L. Using the Chinese herb Scutellaria barbata against extensively drug-resistant Acinetobacter baumannii infections: In vitro and in vivo studies. BMC Complement. Altern. Med. 2018, 18, 96. [Google Scholar] [CrossRef] [PubMed]
  18. Yu, J.; Lei, J.; Yu, H.; Cai, X.; Zou, G. Chemical composition and antimicrobial activity of the essential oil of Scutellaria barbata. Phytochemistry 2004, 65, 881–884. [Google Scholar] [CrossRef] [PubMed]
  19. Sato, Y.; Suzaki, S.; Nishikawa, T.; Kihara, M.; Shibata, H.; Higuti, T. Phytochemical flavones isolated from Scutellaria barbata and antibacterial activity against methicillin-resistant Staphylococcus aureus. J. Ethnopharmacol. 2000, 72, 483–488. [Google Scholar] [CrossRef]
  20. Mei, X.-Y.; Zhou, L.-Y.; Zhang, T.-Y.; Lu, B.; Ji, L.-L. Scutellaria barbata attenuates diabetic retinopathy by preventing retinal inflammation and the decreased expression of tight junction protein. Int. J. Ophthalmol. 2017, 10, 870–877. [Google Scholar]
  21. Wu, X.G.; Wang, S.S.; Miao, H.; Cheng, J.J.; Zhang, S.F.; Shang, Y.Z. Scutellaria barbata flavonoids alleviate memory deficits and neuronal injuries induced by composited Aβ in rats. Behav. Brain Funct. 2016, 12, 33. [Google Scholar] [CrossRef] [Green Version]
  22. Zhang, Y.; Li, Y. Effect of total flavonoids of Scutellaria barbata on cognitive function and nogo-A expression in the hippocampus in cerebral ischemia model in gerbils. Pak. J. Pharm. Sci. 2016, 29, 2373–2376. [Google Scholar]
  23. Wang, Z.; Yu, J.; Wu, J.; Qi, F.; Wang, H.; Wang, Z.; Xu, Z. Scutellarin protects cardiomyocyte ischemia–reperfusion injury by reducing apoptosis and oxidative stress. Life Sci. 2016, 157, 200–207. [Google Scholar] [CrossRef]
  24. Kupferschmidt, K.; Cohen, J. Race to find COVID-19 treatments accelerates. Science 2020, 367, 1412–1413. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  25. Mollica, V.; Rizzo, A.; Massari, F. The pivotal role of TMPRSS2 in coronavirus disease 2019 and prostate cancer. Future Oncol. 2020, 16, 2029–2033. [Google Scholar] [CrossRef] [PubMed]
  26. García-Perdomo, H.A.; Chaves, M.J.; Osorio, J.C.; Sanchez, A. Association between TMPRSS2:ERG fusion gene and the prostate cancer: Systematic review and meta-analysis. Cent. Eur. J. Urol. 2018, 71, 410–419. [Google Scholar]
  27. Kuba, K.; Imai, Y.; Rao, S.; Gao, H.; Guo, F.; Guan, B.; Huan, Y.; Yang, P.; Zhang, Y.; Deng, W.; et al. A crucial role of angiotensin converting enzyme 2 (ACE2) in SARS coronavirus-induced lung injury. Nat. Med. 2005, 11, 875–879. [Google Scholar] [CrossRef] [PubMed]
  28. Stopsack, K.H.; Mucci, L.A.; Antonarakis, E.S.; Nelson, P.S.; Kantoff, P.W. TMPRSS2 and COVID-19: Serendipity or opportunity for intervention? Cancer Discov. 2020, 10, 779–782. [Google Scholar] [CrossRef] [Green Version]
  29. Rahman, N.; Basharat, Z.; Yousuf, M.; Castaldo, G.; Rastrelli, L.; Khan, H. virtual screening of natural products against type II Transmembrane Serine Protease (TMPRSS2), the priming agent of Coronavirus 2 (SARS-CoV-2). Molecules 2020, 25, 2271. [Google Scholar] [CrossRef]
  30. Senthil Kumar, K.J.; Gokila Vani, M.; Wang, C.S.; Chen, C.C.; Chen, Y.C.; Lu, L.P.; Huang, C.H.; Lai, C.S.; Wang, S.Y. Geranium and lemon essential oils and their active compounds downregulate angiotensin-converting enzyme 2 (ACE2), a SARS-CoV-2 spike receptor-binding domain, in epithelial cells. Plants 2020, 9, 770. [Google Scholar] [CrossRef]
  31. Gyebi, G.A.; Adegunloye, A.P.; Ibrahim, I.M.; Ogunyemi, O.M.; Afolabi, S.O.; Ogunro, O.B. Prevention of SARS-CoV-2 cell entry: Insight from in silico interaction of drug-like alkaloids with spike glycoprotein, human ACE2, and TMPRSS2. J. Biomol. Struct. Dyn. 2020, 1–25. [Google Scholar] [CrossRef]
  32. Wang, S.C.; Chen, Y.; Wang, Y.C.; Wang, W.J.; Yang, C.S.; Tsai, C.L.; Hou, M.H.; Chen, H.F.; Shen, Y.C.; Hung, M.C. Tannic acid suppresses SARS-CoV-2 as a dual inhibitor of the viral main protease and the cellular TMPRSS2 protease. Am. J. Cancer Res. 2020, 10, 2535–2545. [Google Scholar]
  33. Zhang, L.; Lin, D.; Sun, X.; Curth, U.; Drosten, C.; Sauerhering, L.; Becker, S.; Rox, K.; Hilgenfeld, R. Crystal structure of SARS-CoV-2 main protease provides a basis for design of improved α-ketoamide inhibitors. Science 2020, 368, 409–412. [Google Scholar] [CrossRef] [Green Version]
  34. Jin, Z.; Du, X.; Xu, Y.; Deng, Y.; Liu, M.; Zhao, Y.; Zhang, B.; Li, X.; Zhang, L.; Peng, C.; et al. Structure of M(pro) from SARS-CoV-2 and discovery of its inhibitors. Nature 2020, 582, 289–293. [Google Scholar] [CrossRef] [Green Version]
  35. Rut, W.; Groborz, K.; Zhang, L.; Sun, X.; Zmudzinski, M.; Pawlik, B.; Wang, X.; Jochmans, D.; Neyts, J.; Młynarski, W.; et al. SARS-CoV-2 M(pro) inhibitors and activity-based probes for patient-sample imaging. Nat. Chem. Biol. 2021, 17, 222–228. [Google Scholar] [CrossRef] [PubMed]
  36. Narkhede, R.R.; Pise, A.V.; Cheke, R.S.; Shinde, S.D. Recognition of natural products as potential inhibitors of COVID-19 main protease (Mpro): In-silico evidences. Nat. Prod. Bioprospect. 2020, 10, 297–306. [Google Scholar] [CrossRef] [PubMed]
  37. Ghosh, R.; Chakraborty, A.; Biswas, A.; Chowdhuri, S. Evaluation of green tea polyphenols as novel corona virus (SARS CoV-2) main protease (Mpro) inhibitors—An in silico docking and molecular dynamics simulation study. J. Biomol. Struct. Dyn. 2020, 22, 1–13. [Google Scholar]
  38. Chowdhury, P. In silico investigation of phytoconstituents from Indian medicinal herb ’Tinospora cordifolia (giloy)’ against SARS-CoV-2 (COVID-19) by molecular dynamics approach. J. Biomol. Struct. Dyn. 2020, 7, 1–18. [Google Scholar] [CrossRef]
  39. Sinha, S.K.; Prasad, S.K.; Islam, M.A.; Chaudhary, S.K.; Singh, S.; Shakya, A. Potential Leads from Liquorice against SARS-CoV-2 Main Protease using Molecular Docking Simulation Studies. Comb. Chem. High Throughput Screen. 2020, 23, 591–597. [Google Scholar]
  40. Yepes-Pérez, A.F.; Herrera-Calderon, O.; Sánchez-Aparicio, J.E.; Tiessler-Sala, L.; Maréchal, J.D.; Cardona, G.W. Investigating potential inhibitory effect of Uncaria tomentosa (Cat’s Claw) against the main protease 3CL(pro) of SARS-CoV-2 by molecular modeling. Evid. Based Complement. Alternat. Med. 2020, 2020, 4932572. [Google Scholar] [CrossRef]
  41. Wang, Y.C.; Yang, W.H.; Yang, C.S.; Hou, M.H.; Tsai, C.L.; Chou, Y.Z.; Hung, M.C.; Chen, Y. Structural basis of SARS-CoV-2 main protease inhibition by a broad-spectrum anti-coronaviral drug. Am. J. Cancer Res. 2020, 10, 2535–2545. [Google Scholar]
  42. Liu, J.; Jiang, M.; Li, Z.; Zhang, X.; Li, X.; Hao, Y.; Su, X.; Zhu, J.; Zheng, C.; Xiao, W.; et al. A Novel systems pharmacology method to investigate molecular mechanisms of Scutellaria barbata D. Don for non-small cell lung cancer. Front. Pharmacol. 2018, 9, 1473. [Google Scholar] [CrossRef] [Green Version]
  43. Li, L.; Xu, X.; Wu, L.; Zhu, H.; He, Z.; Zhang, B.; Chi, Y.; Song, G. Scutellaria barbata polysaccharides inhibit tumor growth and affect the serum proteomic profiling of hepatoma H22-bearing mice. Mol. Med. Rep. 2019, 19, 2254–2262. [Google Scholar] [CrossRef] [Green Version]
  44. Jin, Y.; Chen, W.; Yang, H.; Yan, Z.; Lai, Z.; Feng, J.; Peng, J.; Lin, J. Scutellaria barbata D. Don inhibits migration and invasion of colorectal cancer cells via suppression of PI3K/AKT and TGF-β/Smad signaling pathways. Exp. Ther. Med. 2017, 14, 5527–5534. [Google Scholar] [CrossRef] [Green Version]
  45. Hoffmann, M.; Kleine-Weber, H.; Schroeder, S.; Krüger, N.; Herrler, T.; Erichsen, S.; Schiergens, T.S.; Herrler, G.; Wu, N.H.; Nitsche, A.; et al. SARS-CoV-2 cell entry depends on ACE2 and TMPRSS2 and is blocked by a clinically proven protease inhibitor. Cell 2020, 181, 271–280.e278. [Google Scholar] [CrossRef] [PubMed]
  46. Mi, X.; Zhu, R. Simultaneous determination of 7 active ingredients in Scutellaria barbata D. Don by capillary micellar electrokinetic chromatography. Se Pu Chin. J. Chromatogr. 2010, 28, 209–214. [Google Scholar] [CrossRef] [PubMed]
  47. Goyal, B.; Goyal, D. Targeting the dimerization of the main protease of coronaviruses: A potential broad-spectrum therapeutic strategy. ACS Comb. Sci. 2020, 22, 297–305. [Google Scholar] [CrossRef] [PubMed]
  48. Ma, J.; Huo, X.Q.; Chen, X.; Zhu, W.X.; Yao, M.C.; Qiao, Y.J.; Zhang, Y.L. Study on screening potential traditional Chinese medicines against 2019-nCoV based on Mpro and PLP. Zhongguo Zhong Yao Za Zhi China J. Chin. Materia Med. 2020, 45, 1219–1224. [Google Scholar]
  49. Xia, L.; Shi, Y.; Su, J.; Friedemann, T.; Tao, Z.; Lu, Y.; Ling, Y.; Lv, Y.; Zhao, R.; Geng, Z.; et al. A promising herbal therapy for moderate COVID-19: Antiviral and anti-inflammatory properties, pathways of bioactive compounds, and a clinical real-world pragmatic study. Phytomedicine 2020, 85, 153390. [Google Scholar] [CrossRef] [PubMed]
  50. McKee, D.L.; Sternberg, A.; Stange, U.; Laufer, S.; Naujokat, C. Candidate drugs against SARS-CoV-2 and COVID-19. Pharmacol. Res. 2020, 157, 104859. [Google Scholar] [CrossRef] [PubMed]
  51. Shen, L.W.; Mao, H.J.; Wu, Y.L.; Tanaka, Y.; Zhang, W. TMPRSS2: A potential target for treatment of influenza virus and coronavirus infections. Biochimie 2017, 142, 1–10. [Google Scholar] [CrossRef]
  52. Zhou, Y.; Vedantham, P.; Lu, K.; Agudelo, J.; Carrion, R., Jr.; Nunneley, J.W.; Barnard, D.; Pöhlmann, S.; McKerrow, J.H.; Renslo, A.R.; et al. Protease inhibitors targeting coronavirus and filovirus entry. Antivir. Res. 2015, 116, 76–84. [Google Scholar] [CrossRef]
  53. Chen, Y.; Lear, T.; Evankovich, J.; Larsen, M.; Lin, B.; Alfaras, I.; Kennerdell, J.; Salminen, L.; Camarco, D.; Lockwood, K.; et al. A high throughput screen for TMPRSS2 expression identifies FDA-approved and clinically advanced compounds that can limit SARS-CoV-2 entry. Res. Sq. 2020. [Google Scholar]
  54. Benton, D.J.; Wrobel, A.G.; Xu, P.; Roustan, C.; Martin, S.R.; Rosenthal, P.B.; Skehel, J.J.; Gamblin, S.J. Receptor binding and priming of the spike protein of SARS-CoV-2 for membrane fusion. Nature 2020, 588, 327–330. [Google Scholar] [CrossRef]
  55. Liu, Y.; Jing, Y.Y.; Zeng, C.Y.; Li, C.G.; Xu, L.H.; Yan, L.; Bai, W.J.; Zha, Q.B.; Ouyang, D.Y.; He, X.H. Scutellarin suppresses NLRP3 inflammasome activation in macrophages and protects mice against bacterial sepsis. Front. Pharmacol. 2017, 8, 975. [Google Scholar] [CrossRef] [Green Version]
  56. Sharma, A.; Goyal, S.; Yadav, A.K.; Kumar, P.; Gupta, L. In-silico screening of plant-derived antivirals against main protease, 3CL(pro) and endoribonuclease, NSP15 proteins of SARS-CoV-2. J. Biomol. Struct. Dyn. 2020, 8, 1–15. [Google Scholar]
  57. Bie, B.; Sun, J.; Guo, Y.; Li, J.; Jiang, W.; Yang, J.; Huang, C.; Li, Z. Baicalein: A review of its anti-cancer effects and mechanisms in hepatocellular carcinoma. Biomed. Pharmacother 2017, 93, 1285–1291. [Google Scholar] [CrossRef]
  58. Dinda, B.; Dinda, S.; DasSharma, S.; Banik, R.; Chakraborty, A.; Dinda, M. Therapeutic potentials of baicalin and its aglycone, baicalein against inflammatory disorders. Eur. J. Med. Chem. 2017, 131, 68–80. [Google Scholar] [CrossRef]
  59. Yan, W.; Ma, X.; Zhao, X.; Zhang, S. Baicalein induces apoptosis and autophagy of breast cancer cells via inhibiting PI3K/AKT pathway in vivo and vitro. Drug Des. Dev. Ther. 2018, 12, 3961–3972. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Huang, Y.F.; Bai, C.; He, F.; Xie, Y.; Zhou, H. Review on the potential action mechanisms of Chinese medicines in treating Coronavirus Disease 2019 (COVID-19). Pharmacol. Res. 2020, 158, 104939. [Google Scholar] [CrossRef] [PubMed]
  61. Zhuang, Z.; Wen, J.; Zhang, L.; Zhang, M.; Zhong, X.; Chen, H.; Luo, C. Can network pharmacology identify the anti-virus and anti- inflammatory activities of Shuanghuanglian oral liquid used in Chinese medicine for respiratory tract infection? Eur. J. Integr. Med. 2020, 37, 101139. [Google Scholar] [CrossRef]
  62. Su, H.X.; Yao, S.; Zhao, W.F.; Li, M.J.; Liu, J.; Shang, W.J.; Xie, H.; Ke, C.Q.; Hu, H.C.; Gao, M.N.; et al. Anti-SARS-CoV-2 activities in vitro of Shuanghuanglian preparations and bioactive ingredients. Acta Pharmacol. Sin. 2020, 41, 1167–1177. [Google Scholar] [CrossRef] [PubMed]
  63. Pooja, M.; Reddy, G.J.; Hema, K.; Dodoala, S.; Koganti, B. Unravelling high-affinity binding compounds towards transmembrane protease serine 2 enzyme in treating SARS-CoV-2 infection using molecular modelling and docking studies. Eur. J. Pharmacol. 2021, 890, 173688. [Google Scholar]
  64. Tao, Q.; Du, J.; Li, X.; Zeng, J.; Tan, B.; Xu, J.; Lin, W.; Chen, X.L. Network pharmacology and molecular docking analysis on molecular targets and mechanisms of Huashi Baidu formula in the treatment of COVID-19. Drug Dev. Ind. Pharm. 2020, 46, 1345–1353. [Google Scholar] [CrossRef]
  65. Liu, C.; Zhu, X.; Lu, Y.; Zhang, X.; Jia, X.; Yang, T. Potential treatment of Chinese and Western medicine targeting Nsp14 of SARS-CoV-2. J. Pharm. Anal. 2020, in press. [Google Scholar] [CrossRef]
  66. Nabavi, S.F.; Braidy, N.; Gortzi, O.; Sobarzo-Sanchez, E.; Daglia, M.; Skalicka-Woźniak, K.; Nabavi, S.M. Luteolin as an anti-inflammatory and neuroprotective agent: A brief review. Brain Res. Bull. 2015, 119, 1–11. [Google Scholar] [CrossRef]
  67. Imran, M.; Rauf, A.; Abu-Izneid, T.; Nadeem, M.; Shariati, M.A.; Khan, I.A.; Imran, A.; Orhan, I.E.; Rizwan, M.; Atif, M.; et al. Luteolin, a flavonoid, as an anticancer agent: A review. Biomed. Pharmacother. 2019, 112, 108612. [Google Scholar] [CrossRef]
  68. Ali, F.; Siddique, Y.H. Bioavailability and pharmaco-therapeutic potential of Luteolin in overcoming Alzheimer’s Disease. CNS Neurol. Disord. Drug Targets 2019, 18, 352–365. [Google Scholar] [CrossRef]
  69. Xia, Q.D.; Xun, Y.; Lu, J.L.; Lu, Y.C.; Yang, Y.Y.; Zhou, P.; Hu, J.; Li, C.; Wang, S.G. Network pharmacology and molecular docking analyses on Lianhua Qingwen capsule indicate Akt1 is a potential target to treat and prevent COVID-19. Cell Prolif. 2020, 53, e12949. [Google Scholar] [CrossRef] [PubMed]
  70. Liang, C.; Niu, W.; Wu, F.; Cao, W.; Wu, Z.; Chao, Y.C.; Peng, F. Network pharmacology for the identification of phytochemicals in traditional Chinese medicine for COVID-19 that may regulate interleukin-6. Biosci. Rep. 2021, 41. [Google Scholar] [CrossRef] [PubMed]
  71. Yi, L.; Li, Z.; Yuan, K.; Qu, X.; Chen, J.; Wang, G.; Zhang, H.; Luo, H.; Zhu, L.; Jiang, P.; et al. Small molecules blocking the entry of severe acute respiratory syndrome coronavirus into host cells. J. Virol. 2004, 78, 11334–11339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Ryu, Y.B.; Jeong, H.J.; Kim, J.H.; Kim, Y.M.; Park, J.Y.; Kim, D.; Nguyen, T.T.; Park, S.J.; Chang, J.S.; Park, K.H.; et al. Biflavonoids from Torreya nucifera displaying SARS-CoV 3CL(pro) inhibition. Bioorg. Med. Chem. 2010, 18, 7940–7947. [Google Scholar] [CrossRef]
  73. Maurya, V.K.; Kumar, S.; Prasad, A.K.; Bhatt, M.L.B.; Saxena, S.K. Structure-based drug designing for potential antiviral activity of selected natural products from Ayurveda against SARS-CoV-2 spike glycoprotein and its cellular receptor. Virusdisease 2020, 31, 179–193. [Google Scholar] [CrossRef] [PubMed]
  74. Yu, J.; Liu, H.; Lei, J.; Tan, W.; Hu, X.; Zou, G. Antitumor activity of chloroform fraction of Scutellaria barbata and its active constituents. Phytother. Res. 2007, 21, 817–822. [Google Scholar] [CrossRef] [PubMed]
  75. Du, X.S.; Li, H.D.; Yang, X.J.; Li, J.J.; Xu, J.J.; Chen, Y.; Xu, Q.Q.; Yang, L.; He, C.S.; Huang, C.; et al. Wogonin attenuates liver fibrosis via regulating hepatic stellate cell activation and apoptosis. Int. Immunopharmacol. 2019, 75, 105671. [Google Scholar] [CrossRef] [PubMed]
  76. Palit, P.; Chattopadhyay, D.; Thomas, S.; Kundu, A.; Kim, H.S.; Rezaei, N. Phytopharmaceuticals mediated Furin and TMPRSS2 receptor blocking: Can it be a potential therapeutic option for Covid-19? Phytomedicine 2020, 85, 153396. [Google Scholar] [CrossRef] [PubMed]
  77. Pafumi, I.; Festa, M.; Papacci, F.; Lagostena, L.; Giunta, C.; Gutla, V.; Cornara, L.; Favia, A.; Palombi, F.; Gambale, F.; et al. Naringenin impairs two-pore channel 2 activity and inhibits VEGF-induced angiogenesis. Sci. Rep. 2017, 7, 5121. [Google Scholar] [CrossRef] [PubMed]
  78. Benkerrou, D.; Minicozzi, V.; Gradogna, A.; Milenkovic, S.; Bodrenko, I.V.; Festa, M.; Lagostena, L.; Cornara, L.; D’Amore, A.; Ceccarelli, M.; et al. A perspective on the modulation of plant and animal two pore channels (TPCs) by the flavonoid naringenin. Biophys. Chem. 2019, 254, 106246. [Google Scholar] [CrossRef] [PubMed]
  79. Ou, X.; Liu, Y.; Lei, X.; Li, P.; Mi, D.; Ren, L.; Guo, L.; Guo, R.; Chen, T.; Hu, J.; et al. Characterization of spike glycoprotein of SARS-CoV-2 on virus entry and its immune cross-reactivity with SARS-CoV. Nat. Commun. 2020, 11, 1620. [Google Scholar] [CrossRef] [Green Version]
  80. Nahmias, Y.; Goldwasser, J.; Casali, M.; van Poll, D.; Wakita, T.; Chung, R.T.; Yarmush, M.L. Apolipoprotein B-dependent hepatitis C virus secretion is inhibited by the grapefruit flavonoid naringenin. Hepatology 2008, 47, 1437–1445. [Google Scholar] [CrossRef] [Green Version]
  81. Dong, W.; Wei, X.; Zhang, F.; Hao, J.; Huang, F.; Zhang, C.; Liang, W. A dual character of flavonoids in influenza A virus replication and spread through modulating cell-autonomous immunity by MAPK signaling pathways. Sci. Rep. 2014, 4, 7237. [Google Scholar] [CrossRef]
  82. Cataneo, A.H.D.; Kuczera, D.; Koishi, A.C.; Zanluca, C.; Silveira, G.F.; Arruda, T.B.d.; Suzukawa, A.A.; Bortot, L.O.; Dias-Baruffi, M.; Verri, W.A.; et al. The citrus flavonoid naringenin impairs the in vitro infection of human cells by Zika virus. Sci. Rep. 2019, 9, 16348. [Google Scholar] [CrossRef]
  83. Frabasile, S.; Koishi, A.C.; Kuczera, D.; Silveira, G.F.; Verri, W.A., Jr.; Duarte Dos Santos, C.N.; Bordignon, J. The citrus flavanone naringenin impairs dengue virus replication in human cells. Sci. Rep. 2017, 7, 41864. [Google Scholar] [CrossRef] [Green Version]
  84. Xu, X.; Miao, J.; Shao, Q.; Gao, Y.; Hong, L. Apigenin suppresses influenza A virus-induced RIG-I activation and viral replication. J. Med. Virol. 2020, 92, 3057–3066. [Google Scholar] [CrossRef]
  85. Wu, C.C.; Fang, C.Y.; Cheng, Y.J.; Hsu, H.Y.; Chou, S.P.; Huang, S.Y.; Tsai, C.H.; Chen, J.Y. Inhibition of Epstein-Barr virus reactivation by the flavonoid apigenin. J. Biomed. Sci. 2017, 24, 2. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Qian, S.; Fan, W.; Qian, P.; Zhang, D.; Wei, Y.; Chen, H.; Li, X. Apigenin restricts FMDV infection and inhibits viral IRES driven translational activity. Viruses 2015, 7, 1613–1626. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Hakobyan, A.; Arabyan, E.; Avetisyan, A.; Abroyan, L.; Hakobyan, L.; Zakaryan, H. Apigenin inhibits African swine fever virus infection in vitro. Arch. Virol. 2016, 161, 3445–3453. [Google Scholar] [CrossRef] [PubMed]
  88. Khandelwal, N.; Chander, Y.; Kumar, R.; Riyesh, T.; Dedar, R.K.; Kumar, M.; Gulati, B.R.; Sharma, S.; Tripathi, B.N.; Barua, S.; et al. Antiviral activity of Apigenin against buffalopox: Novel mechanistic insights and drug-resistance considerations. Antivir. Res. 2020, 181, 104870. [Google Scholar] [CrossRef]
  89. Zhang, W.; Qiao, H.; Lv, Y.; Wang, J.; Chen, X.; Hou, Y.; Tan, R.; Li, E. Apigenin inhibits enterovirus-71 infection by disrupting viral RNA association with trans-acting factors. PLoS ONE 2014, 9, e110429. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Scutellaria barbata inhibited the protease activity of SARS-CoV-2 Mpro in vitro FRET-based enzyme activity assay of SARS-CoV-2 Mpro in the absence or presence of Scutellaria barbata treatment with two folds series dilution from 0.5 mg/mL to 4 mg/mL. All data were represented as mean ± standard deviation (SD). Statistical significance was calculated using student t-test. ANOVA was performed when multiple comparisons were evaluated. *, p < 0.05. **, p < 0.01. ***, p < 0.001.
Figure 1. Scutellaria barbata inhibited the protease activity of SARS-CoV-2 Mpro in vitro FRET-based enzyme activity assay of SARS-CoV-2 Mpro in the absence or presence of Scutellaria barbata treatment with two folds series dilution from 0.5 mg/mL to 4 mg/mL. All data were represented as mean ± standard deviation (SD). Statistical significance was calculated using student t-test. ANOVA was performed when multiple comparisons were evaluated. *, p < 0.05. **, p < 0.01. ***, p < 0.001.
Viruses 13 00826 g001
Figure 2. Dose-response curve of SB on SARS-CoV-2 Mpro. SB inhibited SARS-CoV-2 Mpro with half-maximal inhibitory concentration (IC50) of 1.27 mg/mL and Hill slope of 2.5.
Figure 2. Dose-response curve of SB on SARS-CoV-2 Mpro. SB inhibited SARS-CoV-2 Mpro with half-maximal inhibitory concentration (IC50) of 1.27 mg/mL and Hill slope of 2.5.
Viruses 13 00826 g002
Figure 3. Scutellaria barbata inhibited the protease activity of TMPRSS2 in vitro FRET-based enzyme activity assay of TMPRSS2 in the absence or presence of 2 mg/mL Scutellaria barbata treatment. All data were represented as mean ± SD. Statistical significance was calculated using student t-test. * p < 0.05.
Figure 3. Scutellaria barbata inhibited the protease activity of TMPRSS2 in vitro FRET-based enzyme activity assay of TMPRSS2 in the absence or presence of 2 mg/mL Scutellaria barbata treatment. All data were represented as mean ± SD. Statistical significance was calculated using student t-test. * p < 0.05.
Viruses 13 00826 g003
Figure 4. Scutellaria barbata inhibited pseudovirus infection through TMPRSS2. Calu3 and VeroE6 cells with and without TMPRSS2 over-expression were pre-treated with Scutellaria barbata and then infected with SARS-CoV-2 spike pseudovirus. After 24 h infection, infection efficiency rate was measured according to luciferase activities. All data were represented as mean ± SD. Statistical significance was calculated using student t-test. ANOVA was performed when multiple comparisons were evaluated. #, p value < 0.05 compared with vehicle control. *, p value < 0.05 indicates between control cells and TMPRSS2 over-expressing cells.
Figure 4. Scutellaria barbata inhibited pseudovirus infection through TMPRSS2. Calu3 and VeroE6 cells with and without TMPRSS2 over-expression were pre-treated with Scutellaria barbata and then infected with SARS-CoV-2 spike pseudovirus. After 24 h infection, infection efficiency rate was measured according to luciferase activities. All data were represented as mean ± SD. Statistical significance was calculated using student t-test. ANOVA was performed when multiple comparisons were evaluated. #, p value < 0.05 compared with vehicle control. *, p value < 0.05 indicates between control cells and TMPRSS2 over-expressing cells.
Viruses 13 00826 g004
Figure 5. The fingerprint of Scutellaria barbata. Aqueous extracts of Scutellaria barbata was analyzed by LC/MS. The main compositions of individual herbal compounds were indicated by arrows based on the exact molecular weight.
Figure 5. The fingerprint of Scutellaria barbata. Aqueous extracts of Scutellaria barbata was analyzed by LC/MS. The main compositions of individual herbal compounds were indicated by arrows based on the exact molecular weight.
Viruses 13 00826 g005
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Huang, S.-T.; Chen, Y.; Chang, W.-C.; Chen, H.-F.; Lai, H.-C.; Lin, Y.-C.; Wang, W.-J.; Wang, Y.-C.; Yang, C.-S.; Wang, S.-C.; et al. Scutellaria barbata D. Don Inhibits the Main Proteases (Mpro and TMPRSS2) of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection. Viruses 2021, 13, 826. https://doi.org/10.3390/v13050826

AMA Style

Huang S-T, Chen Y, Chang W-C, Chen H-F, Lai H-C, Lin Y-C, Wang W-J, Wang Y-C, Yang C-S, Wang S-C, et al. Scutellaria barbata D. Don Inhibits the Main Proteases (Mpro and TMPRSS2) of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection. Viruses. 2021; 13(5):826. https://doi.org/10.3390/v13050826

Chicago/Turabian Style

Huang, Sheng-Teng, Yeh Chen, Wei-Chao Chang, Hsiao-Fan Chen, Hsiang-Chun Lai, Yu-Chun Lin, Wei-Jan Wang, Yu-Chuan Wang, Chia-Shin Yang, Shao-Chun Wang, and et al. 2021. "Scutellaria barbata D. Don Inhibits the Main Proteases (Mpro and TMPRSS2) of Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV-2) Infection" Viruses 13, no. 5: 826. https://doi.org/10.3390/v13050826

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop